1
|
Hu L, Cohen RI, Barroso M, Boustany NN. Comparison of vinculin tension in cellular monolayers and three-dimensional multicellular aggregates. BIOMEDICAL OPTICS EXPRESS 2024; 15:5199-5214. [PMID: 39296399 PMCID: PMC11407257 DOI: 10.1364/boe.529156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/21/2024]
Abstract
Confocal frequency-domain fluorescence lifetime and Förster resonance energy transfer (FRET) microscopy of Chinese hamster ovary (CHO-K1) cells expressing the vinculin tension sensor (VinTS) is used to compare vinculin tension in three-dimensional (3D) multicellular aggregates and 2D cellular monolayers. In both 2D and 3D cultures, the FRET efficiency of VinTS is 5-6% lower than that of VinTL (p < 0.05), a tail-less control which cannot bind actin or paxillin. The difference between VinTS and VinTL FRET efficiency can be mitigated by treatment with the Rho-associated kinase inhibitor Y-27632, demonstrating that VinTS is under tension in both 2D and 3D cultures. However, there is an overall decrease in FRET efficiency of both VinTS and VinTL in the 3D multicellular aggregates compared with the 2D monolayers. Expression of VinTS in 2D and 3D cultures exhibits puncta consistent with cellular adhesions. While paxillin is present at the sites of VinTS expression in the 2D monolayers, it is generally absent from VinTS puncta in the 3D aggregates. The results suggest that VinTS experiences a modified environment in 3D aggregates compared with 2D monolayers and provide a basis for further investigation of molecular tension sensors in 3D tissue models.
Collapse
Affiliation(s)
- Luni Hu
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Nada N Boustany
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
2
|
Wang S, Castro BA, Katz JL, Arrieta V, Najem H, Vazquez-Cervantes GI, Wan H, Olson IE, Hou D, Dapash M, Billingham LK, Chia TY, Wei C, Rashidi A, Platanias LC, McCortney K, Horbinski CM, Stupp R, Zhang P, Ahmed AU, Sonabend AM, Heimberger AB, Lesniak MS, Riviere-Cazaux C, Burns T, Miska J, Fischietti M, Lee-Chang C. B cell-based therapy produces antibodies that inhibit glioblastoma growth. J Clin Invest 2024; 134:e177384. [PMID: 39207859 PMCID: PMC11473152 DOI: 10.1172/jci177384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and malignant brain tumor with limited therapeutic options and a poor prognosis. Despite current treatments, the invasive nature of GBM often leads to recurrence. A promising alternative strategy is to harness the potential of the immune system against tumor cells. Our previous data showed that the BVax (B cell-based vaccine) can induce therapeutic responses in preclinical models of GBM. In this study, we aimed to characterize the antigenic reactivity of BVax-derived Abs and evaluate their therapeutic potential. We performed immunoproteomics and functional assays in murine models and samples from patients with GBM. Our investigations revealed that BVax distributed throughout the GBM tumor microenvironment and then differentiated into Ab-producing plasmablasts. Proteomics analyses indicated that the Abs produced by BVax had unique reactivity, predominantly targeting factors associated with cell motility and the extracellular matrix. Crucially, these Abs inhibited critical processes such as GBM cell migration and invasion. These findings provide valuable insights into the therapeutic potential of BVax-derived Abs for patients with GBM, pointing toward a novel direction for GBM immunotherapy.
Collapse
Affiliation(s)
- Si Wang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Brandyn A. Castro
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurological Surgery, University of Chicago Medicine, Chicago, Illinois, USA
| | - Joshua L. Katz
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Victor Arrieta
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Hinda Najem
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Gustavo I. Vazquez-Cervantes
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Hanxiao Wan
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Ian E. Olson
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - David Hou
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark Dapash
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leah K. Billingham
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Tzu-yi Chia
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Chao Wei
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Craig M. Horbinski
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peng Zhang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Atique U. Ahmed
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | | | - Terry Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesotta, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Mariafausta Fischietti
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| |
Collapse
|
3
|
Jyoti Mech D, Suhail Rizvi M. Micromechanics of fibrous scaffolds and their stiffness sensing by cells. Biomed Mater 2024; 19:025035. [PMID: 38290154 DOI: 10.1088/1748-605x/ad2409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/30/2024] [Indexed: 02/01/2024]
Abstract
Mechanical properties of the tissue engineering scaffolds are known to play a crucial role in cell response. Therefore, an understanding of the cell-scaffold interactions is of high importance. Here, we have utilized discrete fiber network model to quantitatively study the micromechanics of fibrous scaffolds with different fiber arrangements and cross-linking densities. We observe that localized forces on the scaffold result in its anisotropic deformation even for isotropic fiber arrangements. We also see an exponential decay of the displacement field with distance from the location of applied force. This nature of the decay allows us to estimate the characteristic length for force transmission in fibrous scaffolds. Furthermore, we also looked at the stiffness sensing of fibrous scaffolds by individual cells and its dependence on the cellular sensing mechanism. For this, we considered two conditions- stress-controlled, and strain-controlled application of forces by a cell. With fixed strain, we find that the stiffness sensed by a cell is proportional to the scaffold's 'macroscopic' elastic modulus. However, under fixed stress application by the cell, the stiffness sensed by the cell also depends on the cell's own stiffness. In fact, the stiffness values for the same scaffold sensed by the stiff and soft cells can differ from each other by an order of magnitude. The insights from this work will help in designing tissue engineering scaffolds for applications where mechanical stimuli are a critical factor.
Collapse
Affiliation(s)
- Dhruba Jyoti Mech
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| | - Mohd Suhail Rizvi
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502284, India
- Computational Biology Research Lab, IIT Hyderabad, Kandi, Sangareddy, Telangana 502284, India
| |
Collapse
|
4
|
Lichtenberg JY, Ramamurthy E, Young AD, Redman TP, Leonard CE, Das SK, Fisher PB, Lemmon CA, Hwang PY. Leader cells mechanically respond to aligned collagen architecture to direct collective migration. PLoS One 2024; 19:e0296153. [PMID: 38165954 PMCID: PMC10760762 DOI: 10.1371/journal.pone.0296153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 12/06/2023] [Indexed: 01/04/2024] Open
Abstract
Leader cells direct collective migration through sensing cues in their microenvironment to determine migration direction. The mechanism by which leader cells sense the mechanical cue of organized matrix architecture culminating in a mechanical response is not well defined. In this study, we investigated the effect of organized collagen matrix fibers on leader cell mechanics and demonstrate that leader cells protrude along aligned fibers resulting in an elongated phenotype of the entire cluster. Further, leader cells show increased mechanical interactions with their nearby matrix compared to follower cells, as evidenced by increased traction forces, increased and larger focal adhesions, and increased expression of integrin-α2. Together our results demonstrate changes in mechanical matrix cues drives changes in leader cell mechanoresponse that is required for directional collective migration. Our findings provide new insights into two fundamental components of carcinogenesis, namely invasion and metastasis.
Collapse
Affiliation(s)
- Jessanne Y. Lichtenberg
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ella Ramamurthy
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Bioengineering, University of California Berkeley, Berkeley, California, United States of America
| | - Anna D. Young
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Trey P. Redman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Corinne E. Leonard
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Swadesh K. Das
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Institute of Molecular Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Institute of Molecular Medicine, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Priscilla Y. Hwang
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia, United States of America
- VCU Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
5
|
Mosier JA, Fabiano ED, Ludolph CM, White AE, Reinhart-King CA. Confinement primes cells for faster migration by polarizing active mitochondria. NANOSCALE ADVANCES 2023; 6:209-220. [PMID: 38125598 PMCID: PMC10729874 DOI: 10.1039/d3na00478c] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 11/07/2023] [Indexed: 12/23/2023]
Abstract
Mechanical cues in the tumor microenvironment interplay with internal cellular processes to control cancer cell migration. Microscale pores present in tumor tissue confer varying degrees of confinement on migrating cells, increasing matrix contact and inducing cytoskeletal rearrangement. Previously, we observed that increased collagen matrix contact significantly increased cell migration speed and cell-induced strains within the matrix. However, the effects of this confinement on future cell migration are not fully understood. Here, we use a collagen microtrack platform to determine the effect of confinement on priming MDA-MB-231 cancer cells for fast migration. We show that migration through a confined track results in increased speed and accumulation of migratory machinery, including actin and active mitochondria, in the front of migrating breast cancer cells. By designing microtracks that allow cells to first navigate a region of high confinement, then a region of low confinement, we assessed whether migration in high confinement changes future migratory behavior. Indeed, cells maintain their speed attained in high confinement even after exiting to a region of low confinement, indicating that cells maintain memory of previous matrix cues to fuel fast migration. Active mitochondria maintain their location at the front of the cell even after cells leave high confinement. Furthermore, knocking out vinculin to disrupt focal adhesions disrupts active mitochondrial localization and disrupts the fast migration seen upon release from confinement. Together, these data suggest that active mitochondrial localization in confinement may facilitate fast migration post-confinement. By better understanding how confinement contributes to future cancer cell migration, we can identify potential therapeutic targets to inhibit breast cancer metastasis.
Collapse
Affiliation(s)
- Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University Nashville TN USA
| | - Emily D Fabiano
- Department of Biomedical Engineering, Vanderbilt University Nashville TN USA
| | - Catherine M Ludolph
- Department of Chemical Engineering, University of Texas at Austin Austin TX USA
| | - Addison E White
- Department of Biomedical Engineering, Vanderbilt University Nashville TN USA
| | | |
Collapse
|
6
|
Chirasani VR, Khan MAI, Malavade JN, Dokholyan NV, Hoffman BD, Campbell SL. Molecular basis and cellular functions of vinculin-actin directional catch bonding. Nat Commun 2023; 14:8300. [PMID: 38097542 PMCID: PMC10721916 DOI: 10.1038/s41467-023-43779-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
The ability of cells and tissues to respond differentially to mechanical forces applied in distinct directions is mediated by the ability of load-bearing proteins to preferentially maintain physical linkages in certain directions. However, the molecular basis and biological consequences of directional force-sensitive binding remain unclear. Vinculin (Vcn) is a load-bearing linker protein that exhibits directional catch bonding due to interactions between the Vcn tail domain (Vt) and filamentous (F)-actin. We developed a computational approach to predict Vcn residues involved in directional catch bonding and produced a set of associated Vcn variants with unaltered Vt structure, actin binding, or phospholipid interactions. Incorporation of the variants did not affect Vcn activation but reduced Vcn loading and altered exchange dynamics, consistent with the loss of directional catch bonding. Expression of Vcn variants perturbed the coordination of subcellular structures and cell migration, establishing key cellular functions for Vcn directional catch bonding.
Collapse
Affiliation(s)
- Venkat R Chirasani
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohammad Ashhar I Khan
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juilee N Malavade
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Chemistry, Penn State College of Medicine, Hershey, PA, USA.
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Cell Biology, Duke University, Durham, NC, USA.
| | - Sharon L Campbell
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Mezher M, Dumbali S, Fenn I, Lamb C, Miller C, Sharmin S, Cabe JI, Bejar-Padilla V, Conway D, Maruthamuthu V. Vinculin is essential for sustaining normal levels of endogenous forces at cell-cell contacts. Biophys J 2023; 122:4518-4527. [PMID: 38350000 PMCID: PMC10719050 DOI: 10.1016/j.bpj.2023.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/11/2023] [Accepted: 10/25/2023] [Indexed: 02/15/2024] Open
Abstract
Transmission of cell-generated (i.e., endogenous) tension at cell-cell contacts is crucial for tissue shape changes during morphogenesis and adult tissue repair in tissues such as epithelia. E-cadherin-based adhesions at cell-cell contacts are the primary means by which endogenous tension is transmitted between cells. The E-cadherin-β-catenin-α-catenin complex mechanically couples to the actin cytoskeleton (and thereby the cell's contractile machinery) both directly and indirectly. However, the key adhesion constituents required for substantial endogenous force transmission at these adhesions in cell-cell contacts are unclear. Due to the role of α-catenin as a mechanotransducer that recruits vinculin at cell-cell contacts, we expected α-catenin to be essential for sustaining normal levels of force transmission. Instead, using the traction force imbalance method to determine the inter-cellular force at a single cell-cell contact between cell pairs, we found that it is vinculin that is essential for sustaining normal levels of endogenous force transmission, with absence of vinculin decreasing the inter-cellular tension by over 50%. Our results constrain the potential mechanical pathways of force transmission at cell-cell contacts and suggest that vinculin can transmit forces at E-cadherin adhesions independent of α-catenin, possibly through β-catenin. Furthermore, we tested the ability of lateral cell-cell contacts to withstand external stretch and found that both vinculin and α-catenin are essential to maintain cell-cell contact stability under external forces.
Collapse
Affiliation(s)
- Mazen Mezher
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Sandeep Dumbali
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Ian Fenn
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Carter Lamb
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Conrad Miller
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Saika Sharmin
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Jolene I Cabe
- Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Vidal Bejar-Padilla
- Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Daniel Conway
- Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Venkat Maruthamuthu
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia.
| |
Collapse
|
8
|
Sarkar A, Niraula G, LeVine D, Zhao Y, Tu Y, Mollaeian K, Ren J, Que L, Wang X. Development of a Ratiometric Tension Sensor Exclusively Responding to Integrin Tension Magnitude in Live Cells. ACS Sens 2023; 8:3701-3712. [PMID: 37738233 PMCID: PMC10788086 DOI: 10.1021/acssensors.3c00606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Integrin tensions are critical for cell mechanotransduction. By converting force to fluorescence, molecular tension sensors image integrin tensions in live cells with a high resolution. However, the fluorescence signal intensity results collectively from integrin tension magnitude, tension dwell time, integrin density, sensor accessibility, and so forth, making it highly challenging to specifically monitor the molecular force level of integrin tensions. Here, a ratiometric tension sensor (RTS) was developed to exclusively monitor the integrin tension magnitude. The RTS consists of two tension-sensing units that are coupled in series and always subject to the same integrin tension. These two units are activated by tension to fluoresce in separate spectra and with different activation rates. The ratio of their activation probabilities, reported by fluorescence ratiometric measurement, is solely determined by the local integrin tension magnitude. RTS responded sensitively to the variation of integrin tension magnitude in platelets and focal adhesions due to different cell plating times, actomyosin inhibition, or vinculin knockout. At last, RTS confirmed that integrin tension magnitude in platelets and focal adhesions decreases monotonically with the substrate rigidity, verifying the rigidity dependence of integrin tensions in live cells and suggesting that integrin tension magnitude could be a key biomechanical factor in cell rigidity sensing.
Collapse
Affiliation(s)
- Anwesha Sarkar
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
| | - Gopal Niraula
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
| | - Dana LeVine
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, Iowa 50011, United States
| | - Yuanchang Zhao
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
| | - Ying Tu
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
| | - Keyvan Mollaeian
- Department of Mechanical Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Juan Ren
- Department of Mechanical Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Long Que
- Department of Electrical and Computer Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Xuefeng Wang
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
- Hoxworth Blood Center, College of Medicine, The University of Cincinnati, Cincinnati, Ohio 45219, United States
| |
Collapse
|
9
|
Giordano F, D'Amico M, Montalto FI, Malivindi R, Chimento A, Conforti FL, Pezzi V, Panno ML, Andò S, De Amicis F. Cdk4 Regulates Glioblastoma Cell Invasion and Stemness and Is Target of a Notch Inhibitor Plus Resveratrol Combined Treatment. Int J Mol Sci 2023; 24:10094. [PMID: 37373242 DOI: 10.3390/ijms241210094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive types of cancer characterized by poor patient outcomes. To date, it is believed that the major cause of its recurrence and chemoresistance is represented by the enrichment of GBM stem cells (GSCs) sustained by the abnormal activation of a number of signaling pathways. In this study, we found that in GBM cells, treatment with low toxicity doses of the γ-secretase inhibitor RO4929097 (GSI), blocking the Notch pathway activity, in combination with resveratrol (RSV) was able to reverse the basal mesenchymal phenotype to an epithelial-like phenotype, affecting invasion and stemness interplay. The mechanism was dependent on cyclin D1 and cyclin-dependent kinase (CDK4), leading to a reduction of paxillin (Pxn) phosphorylation. Consequently, we discovered the reduced interaction of Pxn with vinculin (Vcl), which, during cell migration, transmits the intracellular forces to the extracellular matrix. The exogenous expression of a constitutively active Cdk4 mutant prevented the RSV + GSI inhibitory effects in GBM cell motility/invasion and augmented the expression of stemness-specific markers, as well as the neurosphere sizes/forming abilities in untreated cells. In conclusion, we propose that Cdk4 is an important regulator of GBM stem-like phenotypes and invasive capacity, highlighting how the combined treatment of Notch inhibitors and RSV could be prospectively implemented in the novel therapeutic strategies to target Cdk4 for these aggressive brain tumors.
Collapse
Affiliation(s)
- Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Maria D'Amico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Francesca Ida Montalto
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
10
|
Bowers DT, McCulloch ME, Brown JL. Evaluation of focal adhesion mediated subcellular curvature sensing in response to engineered extracellular matrix. Biointerphases 2023; 18:021004. [PMID: 37019799 PMCID: PMC10079328 DOI: 10.1116/6.0002440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/24/2023] [Accepted: 03/14/2023] [Indexed: 04/07/2023] Open
Abstract
Fibril curvature is bioinstructive to attached cells. Similar to natural healthy tissues, an engineered extracellular matrix can be designed to stimulate cells to adopt desired phenotypes. To take full advantage of the curvature control in biomaterial fabrication methodologies, an understanding of the response to fibril subcellular curvature is required. In this work, we examined morphology, signaling, and function of human cells attached to electrospun nanofibers. We controlled curvature across an order of magnitude using nondegradable poly(methyl methacrylate) (PMMA) attached to a stiff substrate with flat PMMA as a control. Focal adhesion length and the distance of maximum intensity from the geographic center of the vinculin positive focal adhesion both peaked at a fiber curvature of 2.5 μm-1 (both ∼2× the flat surface control). Vinculin experienced slightly less tension when attached to nanofiber substrates. Vinculin expression was also more affected by a subcellular curvature than structural proteins α-tubulin or α-actinin. Among the phosphorylation sites we examined (FAK397, 576/577, 925, and Src416), FAK925 exhibited the most dependance on the nanofiber curvature. A RhoA/ROCK dependance of migration velocity across curvatures combined with an observation of cell membrane wrapping around nanofibers suggested a hybrid of migration modes for cells attached to fibers as has been observed in 3D matrices. Careful selection of nanofiber curvature for regenerative engineering scaffolds and substrates used to study cell biology is required to maximize the potential of these techniques for scientific exploration and ultimately improvement of human health.
Collapse
Affiliation(s)
- Daniel T. Bowers
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Mary Elizabeth McCulloch
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802
| | - Justin L. Brown
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802
| |
Collapse
|
11
|
Essa A, Essa ES, El-deeb SM, Seleem HEM, Al Sahlawi M, Al-Omair OA, Shehab-Eldeen S. Elevated Serum Vinculin in Patients with HBV/HCV-Associated Liver Cirrhosis and Hepatocellular Carcinoma: A Pilot Study. Biologics 2023; 17:23-32. [PMID: 36969330 PMCID: PMC10035354 DOI: 10.2147/btt.s405500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/04/2023] [Indexed: 03/20/2023]
Abstract
Background The stiffness of the extracellular matrix (ECM) controls many cellular processes, such as migration and differentiation. Cells detect stiffness through adhesion structures termed focal adhesions (FAs). Vinculin, an actin-binding FA protein, plays a pivotal role in FA-mediated mechanotransduction. Aim This study aimed to explore the role of vinculin in the development of HBV/HCV-induced hepatocellular carcinoma (HCC). Methods Vinculin levels in a total number of 100 serum samples from patients with HBV/HCV-induced liver cirrhosis and HCC, as well as healthy controls, were analyzed using an enzyme-linked immunosorbent assay (ELISA). Results In patients with HCC and liver cirrhosis, the serum vinculin levels were significantly greater than in controls (503.8±242.2 and 728.4±1044.8 vs 77.7±36.1 respectively, p<0.001). However, results showed no link between serum vinculin and the clinicopathological features of HCC. Conclusion Patients with HBVor HCV-induced liver cirrhosis and HCC have significantly higher serum levels of vinculin than do controls. This might point to a potential role for vinculin in the development of HCC. More research into how this protein affects the development of HCC at the molecular level could lead to better clinical treatments and the development of new molecular therapies.
Collapse
Affiliation(s)
- Abdallah Essa
- Tropical Medicine Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Enas Said Essa
- Clinical Pathology Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | - Sara Mahmoud El-deeb
- Clinical Pathology Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | | | - Muthana Al Sahlawi
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Omar Ahmed Al-Omair
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Somaia Shehab-Eldeen
- Tropical Medicine Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
- Correspondence: Somaia Shehab-Eldeen, Tropical Medicine Department, Faculty of Medicine, Menoufia University, Yassen Abd Al Ghafar Street, Shebin Elkom, Menoufia Governorate, 32511, Egypt, Tel +201117251523, Email
| |
Collapse
|
12
|
Chirasani VR, Khan MAI, Malavade JN, Dokholyan NV, Hoffman BD, Campbell SL. Elucidation of the Molecular Basis and Cellular Functions of Vinculin-Actin Directional Catch Bonding. RESEARCH SQUARE 2023:rs.3.rs-2334490. [PMID: 36711743 PMCID: PMC9882595 DOI: 10.21203/rs.3.rs-2334490/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The ability of cells and tissues to differentially resist or adapt to mechanical forces applied in distinct directions is mediated by the ability of load-bearing proteins to preferentially maintain physical linkages in certain directions. However, the molecular basis and biological consequences of directional force-sensitive binding are unclear. Vinculin (Vcn) is a load-bearing linker protein that exhibits directional catch bonding due to interactions between the Vcn tail domain (Vt) and filamentous (F)-actin. We developed a computational approach to predict Vcn residues involved in directional catch bonding and produced a set of associated Vcn variants with unaltered Vt structure, actin binding, or phospholipid interactions. Incorporation of these variants into Vcn biosensors did not perturb Vcn conformation, but reduced Vcn loading consistent with loss of directional catch bonding. Expression of Vcn variants perturbed the coalignment of FAs and F-actin and directed cell migration, establishing key cellular functions for Vcn directional catch bonding.
Collapse
Affiliation(s)
- Venkat R. Chirasani
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohammad Ashhar I. Khan
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Nikolay V. Dokholyan
- Department of Pharmacology, Department of Biochemistry & Molecular Biology, Department of Chemistry, Penn State College of Medicine, Hershey, PA, USA
| | - Brenton D. Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Sharon L. Campbell
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
13
|
Austin J, Tu Y, Pal K, Wang X. Vinculin transmits high-level integrin tensions that are dispensable for focal adhesion formation. Biophys J 2023; 122:156-167. [PMID: 36352785 PMCID: PMC9822790 DOI: 10.1016/j.bpj.2022.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/08/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
Abstract
Focal adhesions (FAs) transmit force and mediate mechanotransduction between cells and the matrix. Previous studies revealed that integrin-transmitted force is critical to regulate FA formation. As vinculin is a prominent FA protein implicated in integrin tension transmission, this work studies the relation among integrin tensions (force), vinculin (protein), and FA formation (structure) by integrin tension manipulation, force visualization and vinculin knockout (KO). Two DNA-based integrin tension tools are adopted: tension gauge tether (TGT) and integrative tension sensor (ITS), with TGT restricting integrin tensions under a designed Ttol (tension tolerance) value and ITS visualizing integrin tensions above the Ttol value by fluorescence. Results show that large FAs (area >1 μm2) were formed on the TGT surface with Ttol of 54 pN but not on those with lower Ttol values. Time-series analysis of FA formation shows that focal complexes (area <0.5 μm2) appeared on all TGT surfaces 20 min after cell plating, but only matured to large FAs on TGT with Ttol of 54 pN. Next, we tested FA formation in vinculin KO cells on TGT surfaces. Surprisingly, the Ttol value of TGT required for large FA formation is drastically decreased to 23 pN. To explore the cause, we visualized integrin tensions in both wild-type and vinculin KO cells using ITS. The results showed that integrin tensions in FAs of wild-type cells frequently activate ITS with Ttol of 54 pN. With vinculin KO, however, integrin tensions in FAs became lower and unable to activate 54 pN ITS. Force signal intensities of integrin tensions reported by 33 and 43 pN ITS were also significantly reduced with vinculin KO, suggesting that vinculin is essential to transmit high-level integrin tensions and involved in transmitting intermediate-level integrin tensions in FAs. However, the high-level integrin tensions transmitted by vinculin are not required by FA formation.
Collapse
Affiliation(s)
- Jacob Austin
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa
| | - Ying Tu
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa
| | - Kaushik Pal
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa
| | - Xuefeng Wang
- Department of Physics and Astronomy, Iowa State University, Ames, Iowa; Department of Biochemistry, Biophysics and Molecular Biology, Ames, Iowa.
| |
Collapse
|
14
|
Pawluchin A, Galic M. Moving through a changing world: Single cell migration in 2D vs. 3D. Front Cell Dev Biol 2022; 10:1080995. [PMID: 36605722 PMCID: PMC9810339 DOI: 10.3389/fcell.2022.1080995] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Migration of single adherent cells is frequently observed in the developing and adult organism and has been the subject of many studies. Yet, while elegant work has elucidated molecular and mechanical cues affecting motion dynamics on a flat surface, it remains less clear how cells migrate in a 3D setting. In this review, we explore the changing parameters encountered by cells navigating through a 3D microenvironment compared to cells crawling on top of a 2D surface, and how these differences alter subcellular structures required for propulsion. We further discuss how such changes at the micro-scale impact motion pattern at the macro-scale.
Collapse
Affiliation(s)
- Anna Pawluchin
- Institute of Medical Physics and Biophysics, Medical Faculty, University of Münster, Münster, Germany
- Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
- CIM-IMRPS Graduate Program, Münster, Germany
| | - Milos Galic
- Institute of Medical Physics and Biophysics, Medical Faculty, University of Münster, Münster, Germany
- Cells in Motion Interfaculty Centre, University of Münster, Münster, Germany
| |
Collapse
|
15
|
Atherton P, Konstantinou R, Neo SP, Wang E, Balloi E, Ptushkina M, Bennett H, Clark K, Gunaratne J, Critchley D, Barsukov I, Manser E, Ballestrem C. Tensin3 interaction with talin drives the formation of fibronectin-associated fibrillar adhesions. J Biophys Biochem Cytol 2022; 221:213452. [PMID: 36074065 PMCID: PMC9462884 DOI: 10.1083/jcb.202107022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 05/30/2022] [Accepted: 08/05/2022] [Indexed: 12/11/2022] Open
Abstract
The formation of healthy tissue involves continuous remodeling of the extracellular matrix (ECM). Whilst it is known that this requires integrin-associated cell-ECM adhesion sites (CMAs) and actomyosin-mediated forces, the underlying mechanisms remain unclear. Here, we examine how tensin3 contributes to the formation of fibrillar adhesions (FBs) and fibronectin fibrillogenesis. Using BioID mass spectrometry and a mitochondrial targeting assay, we establish that tensin3 associates with the mechanosensors such as talin and vinculin. We show that the talin R11 rod domain binds directly to a helical motif within the central intrinsically disordered region (IDR) of tensin3, whilst vinculin binds indirectly to tensin3 via talin. Using CRISPR knock-out cells in combination with defined tensin3 mutations, we show (i) that tensin3 is critical for the formation of α5β1-integrin FBs and for fibronectin fibrillogenesis, and (ii) the talin/tensin3 interaction drives this process, with vinculin acting to potentiate it.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.,Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rafaella Konstantinou
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.,sGSK Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Suat Peng Neo
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Emily Wang
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Eleonora Balloi
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Marina Ptushkina
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Hayley Bennett
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kath Clark
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Jayantha Gunaratne
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - David Critchley
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Igor Barsukov
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Edward Manser
- sGSK Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| |
Collapse
|
16
|
Amirrah IN, Lokanathan Y, Zulkiflee I, Wee MFMR, Motta A, Fauzi MB. A Comprehensive Review on Collagen Type I Development of Biomaterials for Tissue Engineering: From Biosynthesis to Bioscaffold. Biomedicines 2022; 10:2307. [PMID: 36140407 PMCID: PMC9496548 DOI: 10.3390/biomedicines10092307] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Collagen is the most abundant structural protein found in humans and mammals, particularly in the extracellular matrix (ECM). Its primary function is to hold the body together. The collagen superfamily of proteins includes over 20 types that have been identified. Yet, collagen type I is the major component in many tissues and can be extracted as a natural biomaterial for various medical and biological purposes. Collagen has multiple advantageous characteristics, including varied sources, biocompatibility, sustainability, low immunogenicity, porosity, and biodegradability. As such, collagen-type-I-based bioscaffolds have been widely used in tissue engineering. Biomaterials based on collagen type I can also be modified to improve their functions, such as by crosslinking to strengthen the mechanical property or adding biochemical factors to enhance their biological activity. This review discusses the complexities of collagen type I structure, biosynthesis, sources for collagen derivatives, methods of isolation and purification, physicochemical characteristics, and the current development of collagen-type-I-based scaffolds in tissue engineering applications. The advancement of additional novel tissue engineered bioproducts with refined techniques and continuous biomaterial augmentation is facilitated by understanding the conventional design and application of biomaterials based on collagen type I.
Collapse
Affiliation(s)
- Ibrahim N. Amirrah
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - Izzat Zulkiflee
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| | - M. F. Mohd Razip Wee
- Institute of Microengineering and Nanoelectronics (IMEN), Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Antonella Motta
- Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38122 Trento, Italy
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
17
|
Colasurdo M, Nieves EB, Fernández-Yagüe MA, Franck C, García AJ. Adhesive peptide and polymer density modulate 3D cell traction forces within synthetic hydrogels. Biomaterials 2022; 288:121710. [PMID: 35999082 DOI: 10.1016/j.biomaterials.2022.121710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/27/2022] [Accepted: 07/31/2022] [Indexed: 11/30/2022]
Abstract
Cell-extracellular matrix forces provide pivotal signals regulating diverse physiological and pathological processes. Although mechanobiology has been widely studied in two-dimensional configurations, limited research has been conducted in three-dimensional (3D) systems due to the complex nature of mechanics and cellular behaviors. In this study, we established a platform integrating a well-defined synthetic hydrogel system (PEG-4MAL) with 3D traction force microscopy (TFM) methodologies to evaluate deformation and force responses within synthetic microenvironments, providing insights that are not tractable using biological matrices because of the interdependence of biochemical and biophysical properties and complex mechanics. We dissected the contributions of adhesive peptide density and polymer density, which determines hydrogel stiffness, to 3D force generation for fibroblasts. A critical threshold of adhesive peptide density at a constant matrix elasticity is required for cells to generate 3D forces. Furthermore, matrix displacements and strains decreased with matrix stiffness whereas stresses, and tractions increased with matrix stiffness until reaching constant values at higher stiffness values. Finally, Rho-kinase-dependent contractility and vinculin expression are required to generate significant 3D forces in both collagen and synthetic hydrogels. This research establishes a tunable platform for the study of mechanobiology and provides new insights into how cells sense and transmit forces in 3D.
Collapse
Affiliation(s)
- Mark Colasurdo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Elisa B Nieves
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Marc A Fernández-Yagüe
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Christian Franck
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
18
|
The focal adhesion protein β-parvin controls cardiomyocyte shape and sarcomere assembly in response to mechanical load. Curr Biol 2022; 32:3033-3047.e9. [PMID: 35688156 DOI: 10.1016/j.cub.2022.05.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/12/2022] [Accepted: 05/17/2022] [Indexed: 01/01/2023]
Abstract
Physiological and pathological cardiac stress induced by exercise and hypertension, respectively, increase the hemodynamic load for the heart and trigger specific hypertrophic signals in cardiomyocytes leading to adaptive or maladaptive cardiac hypertrophy responses involving a mechanosensitive remodeling of the contractile cytoskeleton. Integrins sense load and have been implicated in cardiac hypertrophy, but how they discriminate between the two types of cardiac stress and translate mechanical loads into specific cytoskeletal signaling pathways is not clear. Here, we report that the focal adhesion protein β-parvin is highly expressed in cardiomyocytes and facilitates the formation of cell protrusions, the serial assembly of newly synthesized sarcomeres, and the hypertrophic growth of neonatal rat ventricular cardiomyocytes (NRVCs) in vitro. In addition, physiological mechanical loading of NRVCs by either the application of cyclic, uni-axial stretch, or culture on physiologically stiff substrates promotes NRVC elongation in a β-parvin-dependent manner, which is achieved by binding of β-parvin to α/β-PIX, which in turn activates Rac1. Importantly, loss-of-function studies in mice also revealed that β-parvin is essential for the exercise-induced cardiac hypertrophy response in vivo. Our results identify β-parvin as a novel mechano-responsive signaling hub in hypertrophic cardiomyocytes that drives cell elongation in response to physiological mechanical loads.
Collapse
|
19
|
Beunk L, Bakker GJ, van Ens D, Bugter J, Gal F, Svoren M, Friedl P, Wolf K. Actomyosin contractility requirements and reciprocal cell-tissue mechanics for cancer cell invasion through collagen-based channels. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2022; 45:48. [PMID: 35575822 PMCID: PMC9110550 DOI: 10.1140/epje/s10189-022-00182-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/04/2022] [Indexed: 05/09/2023]
Abstract
The interstitial tumor microenvironment is composed of heterogeneously organized collagen-rich porous networks as well as channel-like structures and interfaces which provide both barriers and guidance for invading cells. Tumor cells invading 3D random porous collagen networks depend upon actomyosin contractility to deform and translocate the nucleus, whereas Rho/Rho-associated kinase-dependent contractility is largely dispensable for migration in stiff capillary-like confining microtracks. To investigate whether this dichotomy of actomyosin contractility dependence also applies to physiological, deformable linear collagen environments, we developed nearly barrier-free collagen-scaffold microtracks of varying cross section using two-photon laser ablation. Both very narrow and wide tracks supported single-cell migration by either outward pushing of collagen up to four times when tracks were narrow, or cell pulling on collagen walls down to 50% of the original diameter by traction forces of up to 40 nN when tracks were wide, resulting in track widths optimized to single-cell diameter. Targeting actomyosin contractility by synthetic inhibitors increased cell elongation and nuclear shape change in narrow tracks and abolished cell-mediated deformation of both wide and narrow tracks. Accordingly, migration speeds in all channel widths reduced, with migration rates of around 45-65% of the original speed persisting. Together, the data suggest that cells engage actomyosin contraction to reciprocally adjust both own morphology and linear track width to optimal size for effective cellular locomotion.
Collapse
Affiliation(s)
- Lianne Beunk
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Gert-Jan Bakker
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Diede van Ens
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Jeroen Bugter
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Floris Gal
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Martin Svoren
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
- David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Genomics Center, Utrecht, The Netherlands
| | - Katarina Wolf
- Department of Cell Biology, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
20
|
Sung DC, Chen X, Chen M, Yang J, Schultz S, Babu A, Xu Y, Gao S, Keller TCS, Mericko-Ishizuka P, Lee M, Yang Y, Scallan JP, Kahn ML. VE-cadherin enables trophoblast endovascular invasion and spiral artery remodeling during placental development. eLife 2022; 11:e77241. [PMID: 35486098 PMCID: PMC9106330 DOI: 10.7554/elife.77241] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
During formation of the mammalian placenta, trophoblasts invade the maternal decidua and remodel spiral arteries to bring maternal blood into the placenta. This process, known as endovascular invasion, is thought to involve the adoption of functional characteristics of vascular endothelial cells (ECs) by trophoblasts. The genetic and molecular basis of endovascular invasion remains poorly defined, however, and whether trophoblasts utilize specialized endothelial proteins in an analogous manner to create vascular channels remains untested. Vascular endothelial (VE-)cadherin is a homotypic adhesion protein that is expressed selectively by ECs in which it enables formation of tight vessels and regulation of EC junctions. VE-cadherin is also expressed in invasive trophoblasts and is a prime candidate for a molecular mechanism of endovascular invasion by those cells. Here, we show that VE-cadherin is required for trophoblast migration and endovascular invasion into the maternal decidua in the mouse. VE-cadherin deficiency results in loss of spiral artery remodeling that leads to decreased flow of maternal blood into the placenta, fetal growth restriction, and death. These studies identify a non-endothelial role for VE-cadherin in trophoblasts during placental development and suggest that endothelial proteins may play functionally unique roles in trophoblasts that do not simply mimic those in ECs.
Collapse
Affiliation(s)
- Derek C Sung
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Xiaowen Chen
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Mei Chen
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jisheng Yang
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Susan Schultz
- Department of Radiology, Hospital of the University of PennsylvaniaPhiladelphiaUnited States
| | - Apoorva Babu
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yitian Xu
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Siqi Gao
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - TC Stevenson Keller
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Patricia Mericko-Ishizuka
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Michelle Lee
- University Laboratory Animal Resources, University of PennsylvaniaPhiladelphiaUnited States
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, University of South FloridaTampaUnited States
| | - Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, University of South FloridaTampaUnited States
| | - Mark L Kahn
- Cardiovascular Institute, Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
21
|
Yao L, Brice R, Shippy T. A Protein Composite Neural Scaffold Modulates Astrocyte Migration and Transcriptome Profile. Macromol Biosci 2022; 22:e2100406. [PMID: 35014754 PMCID: PMC9012687 DOI: 10.1002/mabi.202100406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/23/2021] [Indexed: 11/09/2022]
Abstract
Bioscaffold implantation is a promising approach to facilitate the repair and regeneration of wounded neural tissue after injury to the spinal cord or peripheral nerves. However, such bioscaffold grafts currently result in only limited functional recovery. The generation of a neural scaffold using a combination of collagen and glutenin is reported. The conduit material and mechanical properties, as well as its effect on astrocyte behavior is tested. After neural injuries, astrocytes move into the lesion and participate in the process of remodeling the micro-architecture of the wounded neural tissue. In this study, human astrocytes grown on glutenin-collagen scaffolds show higher motility and a lower proliferation rate compared with those grown on collagen scaffolds. RNA sequencing reveals that astrocytes grown on the two types of scaffolds show differentially expressed genes in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways such as actin cytoskeleton and focal adhesion that regulate astrocyte migration on scaffolds. The gene expression of aggrecan and versican, chondroitin sulfate proteoglycans that inhibit axonal growth, is down-regulated in astrocytes grown on glutenin-collagen scaffolds. These outcomes indicate that the implantation of glutenin-collagen scaffolds may promote astrocyte function in the neural regeneration process by enhanced cell migration and reduced glial scar formation.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Ryan Brice
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Teresa Shippy
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| |
Collapse
|
22
|
The C-terminal actin-binding domain of talin forms an asymmetric catch bond with F-actin. Proc Natl Acad Sci U S A 2022; 119:e2109329119. [PMID: 35245171 PMCID: PMC8915792 DOI: 10.1073/pnas.2109329119] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Talin is a mechanosensitive adaptor protein that links integrins to the actin cytoskeleton at cell–extracellular matrix adhesions. Although the C-terminal actin-binding domain ABS3 of talin is required for function, it binds weakly to actin in solution. We show that ABS3 binds actin strongly only when subjected to mechanical forces comparable to those generated by the cytoskeleton. Moreover, the interaction between ABS3 and actin depends strongly on the direction of force in a manner predicted to organize actin to facilitate adhesion growth and efficient cytoskeletal force generation. These characteristics can explain how force sensing by talin helps to nucleate adhesions precisely when and where they are required to transmit force between the cytoskeleton and the extracellular matrix. Focal adhesions (FAs) are large, integrin-based protein complexes that link cells to the extracellular matrix (ECM). FAs form only when and where they are necessary to transmit force between the cellular cytoskeleton and the ECM, but how this occurs remains poorly understood. Talin is a 270-kDa adaptor protein that links integrins to filamentous (F)-actin and recruits additional components during FA assembly in a force-dependent manner. Cell biological and developmental data demonstrate that the third and C-terminal F-actin–binding site (ABS3) of talin is required for normal FA formation. However, purified ABS3 binds F-actin only weakly in solution. We used a single molecule optical trap assay to examine how and whether ABS3 binds F-actin under physiologically relevant mechanical loads. We find that ABS3 forms a catch bond with F-actin when force is applied toward the pointed end of the actin filament, with binding lifetimes >100-fold longer than when force is applied toward the barbed end. Long-lived bonds to F-actin under load require the ABS3 C-terminal dimerization domain, whose cleavage has been reported to regulate FA turnover. Our results support a mechanism in which talin ABS3 preferentially binds to and orients actin filaments with barbed ends facing the cell periphery, thus nucleating long-range order in the actin cytoskeleton. We suggest that talin ABS3 may function as a molecular AND gate that allows FA growth only when sufficient integrin density, F-actin polarization, and mechanical tension are simultaneously present.
Collapse
|
23
|
Complete Model of Vinculin Suggests the Mechanism of Activation by Helical Super-Bundle Unfurling. Protein J 2022; 41:55-70. [PMID: 35006498 DOI: 10.1007/s10930-022-10040-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2022] [Indexed: 12/24/2022]
Abstract
To shed light onto the activation mechanism of vinculin, we carried out a detailed refinement of chicken vinculin and compared it to the human protein which is greater than 95% identical. Refinement resulted in a complete and significantly improved model. This model includes important elements such as a pro-rich strap region (PRR) and C-terminus. The conformation of the PRR stabilized by its inter- and intra-molecular contacts shows a dynamic, but relatively stable motif that constitutes a docking platform for multiple molecules. The contact of the C-terminus with the PRR suggests that phosphorylation of Tyr1065 might control activation and membrane binding. Improved electron densities showed the presence of large solvent molecules such as phosphates/sulfates and a head-group of PIP2. The improved model allowed for a computational stability analysis to be performed by the program Corex/Best which located numerous hot-spots of increased and decreased stability. Proximity of the identified binding sites for regulatory partners involved in inducing or suppressing the activation of vinculin to the unstable elements sheds new light onto the activation pathway and differential activation. This stability analysis suggests that the activation pathway proceeds by unfurling of the super-bundle built from four bundles of helices without separation of the Vt region (840-1066) from the head. According to our mechanism, when activating proteins bind at the strap region a separation of N and C terminal bundles occurs, followed by unfurling of the super-bundle and flattening of the general shape of the molecule, which exposes the interaction sites for binding of auxiliary proteins.
Collapse
|
24
|
Hsieh TH, Hsu CY, Yang PJ, Chiu CC, Liang SS, Ou-Yang F, Kan JY, Hou MF, Wang TN, Tsai EM. DEHP mediates drug resistance by directly targeting AhR in human breast cancer. Biomed Pharmacother 2021; 145:112400. [PMID: 34801851 DOI: 10.1016/j.biopha.2021.112400] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 01/25/2023] Open
Abstract
Resistance to chemotherapy and hormonal therapy is a major clinical problem in breast cancer medicine, especially for cancer metastasis and recurrence. Di(2-ethylhexyl)phthalate (DEHP) affects drug resistance by an unknown mechanism of action. Here we analyzed breast cancer patients (N = 457) and found that Σ4MEHP (the sum of MEHP, MEHHP, MECPP and MEOHP concentrations) in urine was significantly higher (P = 0.018) in the recurrent breast cancer group compared with non-recurrent patients. Σ4MEHP-High was positively and significantly correlated with tumor stage (P = 0.005), lymph node status (P = 0.001), estrogen receptor status (P = 0.010), Her2/Neu status (P = 0.004), recurrence (P = 0.000) and tumor size (P = 0.002), as well as an independent prognostic marker (OR = 1.868; 95% CI = 1.424-2.451; P < 0.000) associated with poor survival rates based on a positive Her2/Neu status (P = 0.035). In addition, we found that DEHP inhibited paclitaxel and doxorubicin effects in breast cancer cell lines MCF-7 and MDA-MB-231 and in zebrafish and mouse tumor initiation models. DEHP induced trefoil factor 3 (TFF3) expression through the vinculin/aryl hydrocarbon receptor (AhR)/ERK signaling pathway and induced CYP2D6, CYP2C8 and CYP3A4 expression through the AhR genomic pathway to increase the epithelial-mesenchymal transition (EMT) and doxorubicin metabolism, respectably. DEHP mediated AhR-related alterations in estrogen receptor expression through the ubiquitination system, which decreased tamoxifen effects in AhR knockout mice. These findings suggest a novel therapeutic avenue by targeting AhR in drug-resistant and recurrent breast cancer.
Collapse
Affiliation(s)
- Tsung-Hua Hsieh
- Department of Medical Research, E-Da Hospital/E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chia-Yi Hsu
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Jing Yang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Shin Liang
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fu Ou-Yang
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jung-Yu Kan
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
25
|
Alcantara MC, Suzuki K, Acebedo AR, Sakamoto Y, Nishita M, Minami Y, Kikuchi A, Yamada G. Stage-dependent function of Wnt5a during male external genitalia development. Congenit Anom (Kyoto) 2021; 61:212-219. [PMID: 34255394 DOI: 10.1111/cga.12438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/27/2022]
Abstract
External genitalia development in mice involves multiple developmental processes under the regulation of various signaling pathways. Wnt5a, one of the major Wnt ligands, is a crucial developmental regulator of outgrowing organs such as the limb, the mandible, and the external genitalia. Defects in Wnt5a signaling have been linked to Robinow syndrome, a genetic disorder in which male patients manifest a micropenis and defective urethral tube formation. Whereas Wnt5a is required for cell proliferation during embryonic external genitalia outgrowth, its role for urethral tube formation has yet to be understood. Here, we show that Wnt5a contributes to urethral tube formation as well as external genitalia outgrowth. Wnt5a is expressed in the embryonic external genitalia mesenchyme, and mesenchymal-specific conditional Wnt5a knockout mice resulted in hypospadias-like urethral defects. Early deletion of Wnt5a at E10.5 showed severe defects in both external genitalia outgrowth and urethral tube formation, along with reduced cell proliferation. The severe urethral tube defect persisted during later timing deletion of Wnt5a (E13.5). Further analyses revealed that loss of Wnt5a disrupted cell polarity and led to a reduction of the phosphorylated myosin light chain and the focal adhesion protein, vinculin. Altogether, these results suggest that Wnt5a coordinates cell proliferation and directed cell migration in a stage-dependent manner during male external genitalia development. Furthermore, Wnt5a may regulate cell polarity, focal adhesion formation, and cell contractility, leading to directed cell migration during male-type urethral formation in a manner that has not been reported in other organ fusion events.
Collapse
Affiliation(s)
- Mellissa C Alcantara
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Alvin R Acebedo
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuki Sakamoto
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Michiru Nishita
- Department of Biochemistry, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Yasuhiro Minami
- Faculty of Medical Sciences, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
26
|
Hernández-Bule ML, Toledano-Macías E, Naranjo A, de Andrés-Zamora M, Úbeda A. In vitro stimulation with radiofrequency currents promotes proliferation and migration in human keratinocytes and fibroblasts. Electromagn Biol Med 2021; 40:338-352. [PMID: 34315307 DOI: 10.1080/15368378.2021.1938113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Capacitive-resistive electric transfer (CRET) therapies have been proposed as strategies for regeneration of cutaneous tissue lesions. Previous studies by our group have shown that intermittent stimulation with 448 kHz CRET currents at subthermal densities promotes in vitro proliferation of human stem cells involved in tissue regeneration. The present study investigates the effects of the in vitro exposure to these radiofrequency (RF) currents on the proliferation and migration of keratinocytes and fibroblasts, the main cell types involved in skin regeneration. The effects of the electric stimulation on cell proliferation and migration were studied through XTT and wound closure assays, respectively. The CRET effects on the expression and location of proteins involved in proliferation and migration were assessed by immunoblot and immunofluorescence. The obtained results reveal that electrostimulation promotes proliferation and/or migration in keratinocytes and fibroblasts. These effects would be mediated by changes observed in the expression and location of intercellular adhesion proteins such as β-catenin and E-cadherin, of proteins involved in cell-to-substrate adhesion such as vinculin, p-FAK and the metalloproteinase MMP-9, and of other proteins that control both processes: MAP kinases p-p38, p-JUNK and p-ERK1/2. These responses could represent a mechanism underlying the promotion of normotrophic wound regeneration induced by CRET. Indeed, electric stimulation would favor completion of granulation tissue formation prior to the closure of the outer tissue layers, thus preventing abnormal wound cicatrization or chronification.
Collapse
Affiliation(s)
| | - Elena Toledano-Macías
- Servicio de Bioelectromagnetismo, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Aida Naranjo
- Departamento de Ingeniería Eléctrica, Escuela Técnica Superior de Ingeniería y Diseño Industrial, Universidad Politécnica de Madrid, Madrid, Spain
| | - Marina de Andrés-Zamora
- Departamento de Ingeniería Eléctrica, Escuela Técnica Superior de Ingeniería y Diseño Industrial, Universidad Politécnica de Madrid, Madrid, Spain
| | - Alejandro Úbeda
- Servicio de Bioelectromagnetismo, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| |
Collapse
|
27
|
Bioinspired peptide adhesion on Ti implants alleviates wear particle-induced inflammation and improves interfacial osteogenesis. J Colloid Interface Sci 2021; 605:410-424. [PMID: 34332414 DOI: 10.1016/j.jcis.2021.07.079] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/02/2021] [Accepted: 07/14/2021] [Indexed: 01/18/2023]
Abstract
In the inflammatory peri-implant microenvironment, excessive polarization of macrophages to the proinflammatory M1 phenotype can trigger the secretion of inflammatory cytokines, which promote bone resorption and impede osteogenesis around implants. The direct consequence of this process is the failure of prosthetic implants due to aseptic loosening. To reverse the inflammatory microenvironment and prevent prosthesis loosening, a mussel adhesion-inspired surface strategy was used for bioengineering of titanium implants with integrin-binding ability. In our design, a mussel-inspired catecholic peptide with tetravalent 3,4-dihydroxy-l-phenylalanine (DOPA) and Arg-Gly-Asp (RGD) sequences was synthesized. The peptide can easily anchor to the surface of medical titanium materials through a mussel adhesive mechanism. We found that peptide-decorated titanium implants could effectively inhibit peri-implant inflammation in a wear particle model and could promote the polarization of macrophages to a pro-healing M2 phenotype by interfering with integrin-α2β1 and integrin-αvβ3. Moreover, the peptide coating increased the adherence of osteoblasts and promoted osteogenesis on titanium implants even under inflammatory conditions. This work suggested that this biomimetic catecholic integrin-binding peptide can provide facile tactics for surface bioengineering of medical prostheses with improved interfacial osteogenesis under inflammatory conditions, which might contribute greatly to the prevention of prosthesis loosening and the improvement of clinical outcomes.
Collapse
|
28
|
Justino AB, Florentino RM, França A, Filho ACML, Franco RR, Saraiva AL, Fonseca MC, Leite MF, Salmen Espindola F. Alkaloid and acetogenin-rich fraction from Annona crassiflora fruit peel inhibits proliferation and migration of human liver cancer HepG2 cells. PLoS One 2021; 16:e0250394. [PMID: 34237060 PMCID: PMC8266062 DOI: 10.1371/journal.pone.0250394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/24/2021] [Indexed: 01/09/2023] Open
Abstract
Plant species from Annonaceae are commonly used in traditional medicine to treat various cancer types. This study aimed to investigate the antiproliferative potential of an alkaloid and acetogenin-rich fraction from the fruit peel of Annona crassiflora in HepG2 cells. A liquid-liquid fractionation was carried out on the ethanol extract of A. crassiflora fruit peel in order to obtain an alkaloid and acetogenin-rich fraction (AF-Ac). Cytotoxicity, proliferation and migration were evaluated in the HepG2 cells, as well as the proliferating cell nuclear antigen (PCNA), vinculin and epidermal growth factor receptor (EGFR) expression. In addition, intracellular Ca2+ was determined using Fluo4-AM and fluorescence microscopy. First, 9 aporphine alkaloids and 4 acetogenins that had not yet been identified in the fruit peel of A. crassiflora were found in AF-Ac. The treatment with 50 μg/mL AF-Ac reduced HepG2 cell viability, proliferation and migration (p < 0.001), which is in accordance with the reduced expression of PCNA and EGFR levels (p < 0.05). Furthermore, AF-Ac increased intracellular Ca2+ in the HepG2 cells, mobilizing intracellular calcium stores, which might be involved in the anti-migration and anti-proliferation capacities of AF-Ac. Our results support the growth-inhibitory potential of AF-Ac on HepG2 cells and suggest that this effect is triggered, at least in part, by PCNA and EGFR modulation and mobilization of intracellular Ca2+. This study showed biological activities not yet described for A. crassiflora fruit peel, which provide new possibilities for further in vivo studies to assess the antitumoral potential of A. crassiflora, especially its fruit peel.
Collapse
Affiliation(s)
- Allisson B. Justino
- Institute of Biotechnology, Federal University of Uberlandia, Uberlândia, Minas Gerais, Brazil
| | - Rodrigo M. Florentino
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andressa França
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Molecular Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Antonio C. M. L. Filho
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo R. Franco
- Institute of Biotechnology, Federal University of Uberlandia, Uberlândia, Minas Gerais, Brazil
| | - André L. Saraiva
- Institute of Biotechnology, Federal University of Uberlandia, Uberlândia, Minas Gerais, Brazil
| | - Matheus C. Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Maria F. Leite
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Foued Salmen Espindola
- Institute of Biotechnology, Federal University of Uberlandia, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
29
|
Alshehri S, Susapto HH, Hauser CAE. Scaffolds from Self-Assembling Tetrapeptides Support 3D Spreading, Osteogenic Differentiation, and Angiogenesis of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:2094-2106. [PMID: 33908763 PMCID: PMC8382244 DOI: 10.1021/acs.biomac.1c00205] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/15/2021] [Indexed: 01/01/2023]
Abstract
The apparent rise of bone disorders demands advanced treatment protocols involving tissue engineering. Here, we describe self-assembling tetrapeptide scaffolds for the growth and osteogenic differentiation of human mesenchymal stem cells (hMSCs). The rationally designed peptides are synthetic amphiphilic self-assembling peptides composed of four amino acids that are nontoxic. These tetrapeptides can quickly solidify to nanofibrous hydrogels that resemble the extracellular matrix and provide a three-dimensional (3D) environment for cells with suitable mechanical properties. Furthermore, we can easily tune the stiffness of these peptide hydrogels by just increasing the peptide concentration, thus providing a wide range of peptide hydrogels with different stiffnesses for 3D cell culture applications. Since successful bone regeneration requires both osteogenesis and vascularization, our scaffold was found to be able to promote angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro. The results presented suggest that ultrashort peptide hydrogels are promising candidates for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Salwa Alshehri
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Hepi H. Susapto
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
30
|
Du X, Kong J, Liu Y, Xu Q, Wang K, Huang D, Wei Y, Chen W, Mao H. The Measurement and Analysis of Impedance Response of HeLa Cells to Distinct Chemotherapy Drugs. MICROMACHINES 2021; 12:mi12020202. [PMID: 33669372 PMCID: PMC7920318 DOI: 10.3390/mi12020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/03/2021] [Accepted: 02/13/2021] [Indexed: 12/01/2022]
Abstract
Electric cell–substrate impedance sensing exhibits a real-time and label-free feature to monitor the response of cells stimulated by various biochemical and mechanical signals. Alterations in the currents passing through the cell–electrode system characterize the impedance variations of cells. The impedance responses of HeLa cells under distinct chemotherapy drugs combine the effects of cell proliferation and cell–substrate adhesion. Optimal interdigitated electrodes were selected to explore the impedance responses of HeLa cells. Measurements of impedance of cells in response to three widely used chemotherapy drugs in clinical practice, namely cisplatin, doxorubicin, 5-fluorouracil, were performed. The results demonstrated that distinct impedance responses of HeLa cells to drugs were exhibited and a decrease in measured impedance was observed after drug treatment, accompanied by alterations in the distribution and intensity of the adhesion-related protein vinculin and the rate of cell proliferation. The link between the impedance profiles of HeLa cells and their biological functions was developed based on the circuit model. This study demonstrated the weights of cell proliferation and adhesion of HeLa cells under the treatments of DDP, DOX, and 5-FU, resulted in distinct impedance responses of cells, providing an impedance-based evaluation methodology for cervical cancer treatment.
Collapse
Affiliation(s)
- Xiangbin Du
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
- Shanxi Key Laboratory of Material Strength & Structural Impact, Institute of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jinlong Kong
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
- Shanxi Key Laboratory of Material Strength & Structural Impact, Institute of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yang Liu
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qianmin Xu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
| | - Kaiqun Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
- Shanxi Key Laboratory of Material Strength & Structural Impact, Institute of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- Correspondence: (K.W.); (H.M.); Tel.: +86-139-3421-2990 (K.W.); +86-158-0125-6264 (H.M.)
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
- Shanxi Key Laboratory of Material Strength & Structural Impact, Institute of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
| | - Weiyi Chen
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China; (X.D.); (J.K.); (Q.X.); (D.H.); (Y.W.); (W.C.)
- Shanxi Key Laboratory of Material Strength & Structural Impact, Institute of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Haiyang Mao
- Institute of Microelectronics of Chinese Academy of Sciences, Beijing 100029, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (K.W.); (H.M.); Tel.: +86-139-3421-2990 (K.W.); +86-158-0125-6264 (H.M.)
| |
Collapse
|
31
|
Manipulation of Focal Adhesion Signaling by Pathogenic Microbes. Int J Mol Sci 2021; 22:ijms22031358. [PMID: 33572997 PMCID: PMC7866387 DOI: 10.3390/ijms22031358] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 12/22/2022] Open
Abstract
Focal adhesions (FAs) serve as dynamic signaling hubs within the cell. They connect intracellular actin to the extracellular matrix (ECM) and respond to environmental cues. In doing so, these structures facilitate important processes such as cell-ECM adhesion and migration. Pathogenic microbes often modify the host cell actin cytoskeleton in their pursuit of an ideal replicative niche or during invasion to facilitate uptake. As actin-interfacing structures, FA dynamics are also intimately tied to actin cytoskeletal organization. Indeed, exploitation of FAs is another avenue by which pathogenic microbes ensure their uptake, survival and dissemination. This is often achieved through the secretion of effector proteins which target specific protein components within the FA. Molecular mimicry of the leucine-aspartic acid (LD) motif or vinculin-binding domains (VBDs) commonly found within FA proteins is a common microbial strategy. Other effectors may induce post-translational modifications to FA proteins through the regulation of phosphorylation sites or proteolytic cleavage. In this review, we present an overview of the regulatory mechanisms governing host cell FAs, and provide examples of how pathogenic microbes have evolved to co-opt them to their own advantage. Recent technological advances pose exciting opportunities for delving deeper into the mechanistic details by which pathogenic microbes modify FAs.
Collapse
|
32
|
Beroz F, Zhou D, Mao X, Lubensky DK. Physical limits to sensing material properties. Nat Commun 2020; 11:5170. [PMID: 33056989 PMCID: PMC7560877 DOI: 10.1038/s41467-020-18995-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/25/2020] [Indexed: 12/03/2022] Open
Abstract
All materials respond heterogeneously at small scales, which limits what a sensor can learn. Although previous studies have characterized measurement noise arising from thermal fluctuations, the limits imposed by structural heterogeneity have remained unclear. In this paper, we find that the least fractional uncertainty with which a sensor can determine a material constant λ0 of an elastic medium is approximately [Formula: see text] for a ≫ d ≫ ξ, [Formula: see text], and D > 1, where a is the size of the sensor, d is its spatial resolution, ξ is the correlation length of fluctuations in λ0, Δλ is the local variability of λ0, and D is the dimension of the medium. Our results reveal how one can construct devices capable of sensing near these limits, e.g. for medical diagnostics. We use our theoretical framework to estimate the limits of mechanosensing in a biopolymer network, a sensory process involved in cellular behavior, medical diagnostics, and material fabrication.
Collapse
Affiliation(s)
- Farzan Beroz
- Department of Physics, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Di Zhou
- Department of Physics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiaoming Mao
- Department of Physics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David K Lubensky
- Department of Physics, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
33
|
Alpha KM, Xu W, Turner CE. Paxillin family of focal adhesion adaptor proteins and regulation of cancer cell invasion. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:1-52. [PMID: 32859368 PMCID: PMC7737098 DOI: 10.1016/bs.ircmb.2020.05.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The paxillin family of proteins, including paxillin, Hic-5, and leupaxin, are focal adhesion adaptor/scaffolding proteins which localize to cell-matrix adhesions and are important in cell adhesion and migration of both normal and cancer cells. Historically, the role of these proteins in regulating the actin cytoskeleton through focal adhesion-mediated signaling has been well documented. However, studies in recent years have revealed additional functions in modulating the microtubule and intermediate filament cytoskeletons to affect diverse processes including cell polarization, vesicle trafficking and mechanosignaling. Expression of paxillin family proteins in stromal cells is also important in regulating tumor cell migration and invasion through non-cell autonomous effects on the extracellular matrix. Both paxillin and Hic-5 can also influence gene expression through a variety of mechanisms, while their own expression is frequently dysregulated in various cancers. Accordingly, these proteins may serve as valuable targets for novel diagnostic and treatment approaches in cancer.
Collapse
Affiliation(s)
- Kyle M Alpha
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weiyi Xu
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Christopher E Turner
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, United States.
| |
Collapse
|
34
|
Vaeyens MM, Jorge-Peñas A, Barrasa-Fano J, Shapeti A, Roeffaers M, Van Oosterwyck H. Actomyosin-dependent invasion of endothelial sprouts in collagen. Cytoskeleton (Hoboken) 2020; 77:261-276. [PMID: 32588525 DOI: 10.1002/cm.21624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/11/2020] [Accepted: 06/22/2020] [Indexed: 12/30/2022]
Abstract
During sprouting angiogenesis-the growth of blood vessels from the existing vasculature-endothelial cells (ECs) adopt an elongated invasive form and exert forces at cell-cell and cell-matrix interaction sites. These cell shape changes and cellular tractions require extensive reorganizations of the actomyosin network. However, the respective roles of actin and myosin for endothelial sprouting are not fully elucidated. In this study, we further investigate these roles by treating 2D-migrating and 3D-sprouting ECs with chemical compounds targeting either myosin or actin. These treatments affected the endothelial cytoskeleton drastically and reduced the invasive response in a compound-specific manner; pointing toward a tight control of the actin and myosin activity during sprouting. Clusters in the data further illustrate that endothelial sprout morphology is sensitive to the in vitro model mechanical microenvironment and directs future research toward mechanical substrate guidance as a strategy for promoting engineered tissue vascularization. In summary, our results add to a growing corpus of research highlighting a key role of the cytoskeleton for sprouting angiogenesis.
Collapse
Affiliation(s)
- Marie-Mo Vaeyens
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Alvaro Jorge-Peñas
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jorge Barrasa-Fano
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Apeksha Shapeti
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Maarten Roeffaers
- Department of Microbial and Molecular Systems (M2S), Centre for Membrane Separations, Adsorption, Catalysis and Spectroscopy for Sustainable Solutions (cMACS), KU Leuven, Leuven, Belgium
| | - Hans Van Oosterwyck
- Biomechanics Section (BMe), Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Sriramulu S, Banerjee A, Jothimani G, Pathak S. Conditioned medium from the human umbilical cord-mesenchymal stem cells stimulate the proliferation of human keratinocytes. J Basic Clin Physiol Pharmacol 2020; 32:51-56. [PMID: 32549126 DOI: 10.1515/jbcpp-2019-0283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 02/08/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES Wound healing is a complex process with a sequence of restoring and inhibition events such as cell proliferation, differentiation, migration as well as adhesion. Mesenchymal stem cells (MSC) derived conditioned medium (CM) has potent therapeutic functions and promotes cell proliferation, anti-oxidant, immunosuppressive, and anti-apoptotic effects. The main aim of this research is to study the role of human umbilical cord-mesenchymal stem cells (UC-MSCs) derived CM in stimulating the proliferation of human keratinocytes (HaCaT). METHODS Firstly, MSC were isolated from human umbilical cords (UC) and the cells were then cultured in proliferative medium. We prepared and collected the CM after 72 h. Morphological changes were observed after the treatment of HaCaT cells with CM. To validate the findings, proliferation rate, clonal efficiency and also gene expression studies were performed. RESULTS Increased proliferation rate was observed and confirmed with the expression of Proliferating Cell Nuclear Antigen (PCNA) after treatment with HaCaT cells. Cell-cell strap formation was also observed when HaCaT cells were treated with CM for a period of 5-6 days which was confirmed by the increased expression of Collagen Type 1 Alpha 1 chain (Col1A1). CONCLUSIONS Our results from present study depicts that the secretory components in the CM might play a significant role by interacting with keratinocytes to promote proliferation and migration. Thus, the CM stimulates cellular proliferation, epithelialization and migration of skin cells which might be the future promising application in wound healing.
Collapse
Affiliation(s)
- Sushmitha Sriramulu
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603103, TN, India
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603103, TN, India
| | - Ganesan Jothimani
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603103, TN, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai, 603103, TN, India
| |
Collapse
|
36
|
Fischer T, Hayn A, Mierke CT. Effect of Nuclear Stiffness on Cell Mechanics and Migration of Human Breast Cancer Cells. Front Cell Dev Biol 2020; 8:393. [PMID: 32548118 PMCID: PMC7272586 DOI: 10.3389/fcell.2020.00393] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
The migration and invasion of cancer cells through 3D confined extracellular matrices is coupled to cell mechanics and the mechanics of the extracellular matrix. Cell mechanics is mainly determined by both the mechanics of the largest organelle in the cell, the nucleus, and the cytoskeletal architecture of the cell. Hence, cytoskeletal and nuclear mechanics are the major contributors to cell mechanics. Among other factors, steric hindrances of the extracellular matrix confinement are supposed to affect nuclear mechanics and thus also influence cell mechanics. Therefore, we propose that the percentage of invasive cells and their invasion depths into loose and dense 3D extracellular matrices is regulated by both nuclear and cytoskeletal mechanics. In order to investigate the effect of both nuclear and cytoskeletal mechanics on the overall cell mechanics, we firstly altered nuclear mechanics by the chromatin de-condensing reagent Trichostatin A (TSA) and secondly altered cytoskeletal mechanics by addition of actin polymerization inhibitor Latrunculin A and the myosin inhibitor Blebbistatin. In fact, we found that TSA-treated MDA-MB-231 human breast cancer cells increased their invasion depth in dense 3D extracellular matrices, whereas the invasion depths in loose matrices were decreased. Similarly, the invasion depths of TSA-treated MCF-7 human breast cancer cells in dense matrices were significantly increased compared to loose matrices, where the invasion depths were decreased. These results are also valid in the presence of a matrix-metalloproteinase inhibitor GM6001. Using atomic force microscopy (AFM), we found that the nuclear stiffnesses of both MDA-MB-231 and MCF-7 breast cancer cells were pronouncedly higher than their cytoskeletal stiffness, whereas the stiffness of the nucleus of human mammary epithelial cells was decreased compared to their cytoskeleton. TSA treatment reduced cytoskeletal and nuclear stiffness of MCF-7 cells, as expected. However, a softening of the nucleus by TSA treatment may induce a stiffening of the cytoskeleton of MDA-MB-231 cells and subsequently an apparent stiffening of the nucleus. Inhibiting actin polymerization using Latrunculin A revealed a softer nucleus of MDA-MB-231 cells under TSA treatment. This indicates that the actin-dependent cytoskeletal stiffness seems to be influenced by the TSA-induced nuclear stiffness changes. Finally, the combined treatment with TSA and Latrunculin A further justifies the hypothesis of apparent nuclear stiffening, indicating that cytoskeletal mechanics seem to be regulated by nuclear mechanics.
Collapse
Affiliation(s)
- Tony Fischer
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Alexander Hayn
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| |
Collapse
|
37
|
Lee AH, Ghosh D, Quach N, Schroeder D, Dawson MR. Ovarian Cancer Exosomes Trigger Differential Biophysical Response in Tumor-Derived Fibroblasts. Sci Rep 2020; 10:8686. [PMID: 32457479 PMCID: PMC7250846 DOI: 10.1038/s41598-020-65628-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Exosomes are cell-secreted microvesicles that play important roles in epithelial ovarian cancer (EOC) progression, as they are constantly secreted into ascites fluids. While cells spontaneously release exosomes, alterations in intracellular calcium or extracellular pH can release additional exosomes. Yet, little is known about how these exosomes compare to those that are continuously released without stimulation and how they mediate cellular activities important in cancer progression. Here, we demonstrate that chelation of extracellular calcium leads to release of chelation-induced exosomes (CI-exosomes) from OVCAR-3 EOC cells. CI-exosomes display a unique miRNA profile compared to naturally secreted exosomes (SEC-exosomes). Furthermore, treatment with CI- and SEC-exosomes leads to differential biophysical and functional changes including, adhesion and migration in EOC-derived fibroblasts that suggest the development of a malignant tumor microenvironment. This result highlights how tumor environmental factors contribute to heterogeneity in exosome populations and how different exosome populations mediate diversity in stromal cell behavior.
Collapse
Affiliation(s)
- Amy H Lee
- Brown University, School of Engineering, Center for Biomedical Engineering, Providence, RI, 02912, USA
| | - Deepraj Ghosh
- Brown University, Department of Molecular Pharmacology, Physiology, and Biotechnology, Providence, RI, 02912, USA
| | - Nhat Quach
- Brown University, Department of Molecular Pharmacology, Physiology, and Biotechnology, Providence, RI, 02912, USA
| | - Devin Schroeder
- Brown University, Department of Molecular Pharmacology, Physiology, and Biotechnology, Providence, RI, 02912, USA
| | - Michelle R Dawson
- Brown University, School of Engineering, Center for Biomedical Engineering, Providence, RI, 02912, USA. .,Brown University, Department of Molecular Pharmacology, Physiology, and Biotechnology, Providence, RI, 02912, USA.
| |
Collapse
|
38
|
Kim DS, Choi YW, Shanmugasundaram A, Jeong YJ, Park J, Oyunbaatar NE, Kim ES, Choi M, Lee DW. Highly durable crack sensor integrated with silicone rubber cantilever for measuring cardiac contractility. Nat Commun 2020; 11:535. [PMID: 31988308 PMCID: PMC6985253 DOI: 10.1038/s41467-019-14019-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023] Open
Abstract
To date, numerous biosensing platforms have been developed for assessing drug-induced cardiac toxicity by measuring the change in contractile force of cardiomyocytes. However, these low sensitivity, low-throughput, and time-consuming processes are severely limited in their real-time applications. Here, we propose a cantilever device integrated with a polydimethylsiloxane (PDMS)-encapsulated crack sensor to measure cardiac contractility. The crack sensor is chemically bonded to a PDMS thin layer that allows it to be operated very stably in culture media. The reliability of the proposed crack sensor has been improved dramatically compared to no encapsulation layer. The highly sensitive crack sensor continuously measures the cardiac contractility without changing its gauge factor for up to 26 days (>5 million heartbeats), while changes in contractile force induced by drugs are monitored using the crack sensor-integrated cantilever. Finally, experimental results are compared with those obtained via conventional optical methods to verify the feasibility of building a contraction-based drug-toxicity testing system. Measuring cardiac contractility is challenging. Here, the authors encapsulated a crack-based sensor with polydimethylsiloxane, thereby endowing the sensor with the stability to measure cardiac contractility for up to 26 days as well as monitoring drug-induced cardiac toxicity in cell culture.
Collapse
Affiliation(s)
- Dong-Su Kim
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yong Whan Choi
- Division of Mechanical Convergence Engineering, College of MICT Convergence Engineering, Silla University, Busan, 46958, Republic of Korea
| | - Arunkumar Shanmugasundaram
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yun-Jin Jeong
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jongsung Park
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Nomin-Erdene Oyunbaatar
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Eung-Sam Kim
- Department of Biological Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.,Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Mansoo Choi
- Global Frontier Center for Multiscale Energy Systems, Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dong-Weon Lee
- MEMS and Nanotechnology Laboratory, School of Mechanical Systems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea. .,Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
39
|
Anguiano M, Morales X, Castilla C, Pena AR, Ederra C, Martínez M, Ariz M, Esparza M, Amaveda H, Mora M, Movilla N, Aznar JMG, Cortés-Domínguez I, Ortiz-de-Solorzano C. The use of mixed collagen-Matrigel matrices of increasing complexity recapitulates the biphasic role of cell adhesion in cancer cell migration: ECM sensing, remodeling and forces at the leading edge of cancer invasion. PLoS One 2020; 15:e0220019. [PMID: 31945053 PMCID: PMC6964905 DOI: 10.1371/journal.pone.0220019] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/02/2020] [Indexed: 11/19/2022] Open
Abstract
The migration of cancer cells is highly regulated by the biomechanical properties of their local microenvironment. Using 3D scaffolds of simple composition, several aspects of cancer cell mechanosensing (signal transduction, EMC remodeling, traction forces) have been separately analyzed in the context of cell migration. However, a combined study of these factors in 3D scaffolds that more closely resemble the complex microenvironment of the cancer ECM is still missing. Here, we present a comprehensive, quantitative analysis of the role of cell-ECM interactions in cancer cell migration within a highly physiological environment consisting of mixed Matrigel-collagen hydrogel scaffolds of increasing complexity that mimic the tumor microenvironment at the leading edge of cancer invasion. We quantitatively show that the presence of Matrigel increases hydrogel stiffness, which promotes β1 integrin expression and metalloproteinase activity in H1299 lung cancer cells. Then, we show that ECM remodeling activity causes matrix alignment and compaction that favors higher tractions exerted by the cells. However, these traction forces do not linearly translate into increased motility due to a biphasic role of cell adhesions in cell migration: at low concentration Matrigel promotes migration-effective tractions exerted through a high number of small sized focal adhesions. However, at high Matrigel concentration, traction forces are exerted through fewer, but larger focal adhesions that favor attachment yielding lower cell motility.
Collapse
Affiliation(s)
- María Anguiano
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Xabier Morales
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Carlos Castilla
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Alejandro Rodríguez Pena
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Cristina Ederra
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Martín Martínez
- Neuroimaging Laboratory, Division of Neurosciences, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Mikel Ariz
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Maider Esparza
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Hippolyte Amaveda
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Mario Mora
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Nieves Movilla
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - José Manuel García Aznar
- Department of Mechanical Engineering, Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
| | - Iván Cortés-Domínguez
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Carlos Ortiz-de-Solorzano
- IDISNA, Ciberonc and Solid Tumours and Biomarkers Program, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- * E-mail:
| |
Collapse
|
40
|
Silvestre JG, Baptista IL, Silva WJ, Cruz A, Silva MT, Miyabara EH, Labeit S, Moriscot AS. The E3 ligase MuRF2 plays a key role in the functional capacity of skeletal muscle fibroblasts. ACTA ACUST UNITED AC 2019; 52:e8551. [PMID: 31482977 PMCID: PMC6720025 DOI: 10.1590/1414-431x20198551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/11/2019] [Indexed: 12/13/2022]
Abstract
Fibroblasts are a highly heterogeneous population of cells, being found in a large number of different tissues. These cells produce the extracellular matrix, which is essential to preserve structural integrity of connective tissues. Fibroblasts are frequently engaged in migration and remodeling, exerting traction forces in the extracellular matrix, which is crucial for matrix deposition and wound healing. In addition, previous studies performed on primary myoblasts suggest that the E3 ligase MuRF2 might function as a cytoskeleton adaptor. Here, we hypothesized that MuRF2 also plays a functional role in skeletal muscle fibroblasts. We found that skeletal muscle fibroblasts express MuRF2 and its siRNA knock-down promoted decreased fibroblast migration, cell border accumulation of polymerized actin, and down-regulation of the phospho-Akt expression. Our results indicated that MuRF2 was necessary to maintain the actin cytoskeleton functionality in skeletal muscle fibroblasts via Akt activity and exerted an important role in extracellular matrix remodeling in the skeletal muscle tissue.
Collapse
Affiliation(s)
- J G Silvestre
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - I L Baptista
- Faculdade de Ciências Aplicadas, UNICAMP, Limeira, SP, Brasil
| | - W J Silva
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - A Cruz
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - M T Silva
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - E H Miyabara
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| | - S Labeit
- Institute for Integrative Pathophysiology, Mannheim Medical University, Faculty for Clinical Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - A S Moriscot
- Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
41
|
Chang LY, Fan SMY, Liao YC, Wang WH, Chen YJ, Lin SJ. Proteomic Analysis Reveals Anti-Fibrotic Effects of Blue Light Photobiomodulation on Fibroblasts. Lasers Surg Med 2019; 52:358-372. [PMID: 31321797 DOI: 10.1002/lsm.23137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND OBJECTIVES This study was aimed at determining the effects of blue light photobiomodulation on primary adult mouse dermal fibroblasts (AMDFs) and the associated signaling pathways. STUDY DESIGN/MATERIALS AND METHODS Cultured AMDFs from adult C57BL/6 mice were irradiated by blue light from a light-emitting diode (wavelength = 463 ± 50 nm; irradiance = 5 mW/cm2 ; energy density = 4-8 J/cm2 ). The cells were analyzed using mass spectrometry for proteomics/phosphoproteomics, AlamarBlue assay for mitochondrial activity, time-lapse video for cell migration, quantitative polymerase chain reaction for gene expression, and immunofluorescence for protein expression. RESULTS Proteomic/phosphoproteomic analysis showed inhibition of extracellular signal-regulated kinases/mammalian target of rapamycin and casein kinase 2 pathways, cell motility-related networks, and multiple metabolic processes, including carbon metabolism, biosynthesis of amino acid, glycolysis/gluconeogenesis, and the pentose phosphate pathway. Functional analysis demonstrated inhibition of mitochondrial activities, cell migration, and mitosis. Expression of growth promoting insulin-like growth factor 1 and fibrosis-related genes, including transforming growth factor β1 (TGFβ1) and collagen type 1 ɑ2 chain diminished. Protein expression of α-smooth muscle actin, an important regulator of myofibroblast functions, was also suppressed. CONCLUSIONS Low-level blue light exerted suppressive effects on AMDFs, including suppression of mitochondrial activity, metabolism, cell motility, proliferation, TGFβ1 levels, and collagen I production. Low-level blue light can be a potential treatment for the prevention and reduction of tissue fibrosis, such as hypertrophic scar and keloids. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lo-Yu Chang
- School of Medicine, College of Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Sabrina Mai-Yi Fan
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yen-Chen Liao
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Wei-Hung Wang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Yu-Ju Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 106, Taiwan.,Institute of Chemistry, Academia Sinica, No. 128, Sec. 2, Academia Rd, Taipei 115, Taiwan
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Chung-Shan South Road, Taipei 100, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 1, Jen-Ai Road, Taipei 100, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei 100, Taiwan
| |
Collapse
|
42
|
Yoon C, Choi C, Stapleton S, Mirabella T, Howes C, Dong L, King J, Yang J, Oberai A, Eyckmans J, Chen CS. Myosin IIA-mediated forces regulate multicellular integrity during vascular sprouting. Mol Biol Cell 2019; 30:1974-1984. [PMID: 31318321 PMCID: PMC6727772 DOI: 10.1091/mbc.e19-02-0076] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Angiogenic sprouting is a critical process involved in vascular network formation within tissues. During sprouting, tip cells and ensuing stalk cells migrate collectively into the extracellular matrix while preserving cell–cell junctions, forming patent structures that support blood flow. Although several signaling pathways have been identified as controlling sprouting, it remains unclear to what extent this process is mechanoregulated. To address this question, we investigated the role of cellular contractility in sprout morphogenesis, using a biomimetic model of angiogenesis. Three-dimensional maps of mechanical deformations generated by sprouts revealed that mainly leader cells, not stalk cells, exert contractile forces on the surrounding matrix. Surprisingly, inhibiting cellular contractility with blebbistatin did not affect the extent of cellular invasion but resulted in cell–cell dissociation primarily between tip and stalk cells. Closer examination of cell–cell junctions revealed that blebbistatin impaired adherens-junction organization, particularly between tip and stalk cells. Using CRISPR/Cas9-mediated gene editing, we further identified NMIIA as the major isoform responsible for regulating multicellularity and cell contractility during sprouting. Together, these studies reveal a critical role for NMIIA-mediated contractile forces in maintaining multicellularity during sprouting and highlight the central role of forces in regulating cell–cell adhesions during collective motility.
Collapse
Affiliation(s)
- Christine Yoon
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Colin Choi
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Sarah Stapleton
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Teodelinda Mirabella
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Caroline Howes
- Department of Mechanical, Aerospace and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Li Dong
- Department of Mechanical, Aerospace and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180.,The Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX 78712
| | - Jessica King
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Jinling Yang
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Assad Oberai
- Department of Mechanical, Aerospace and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180.,Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90007
| | - Jeroen Eyckmans
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215.,Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| |
Collapse
|
43
|
Inactivation of Lgi1 in murine neuronal precursor cells leads to dysregulation of axon guidance pathways. Genomics 2019; 112:1167-1172. [PMID: 31276752 DOI: 10.1016/j.ygeno.2019.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/22/2019] [Accepted: 07/01/2019] [Indexed: 11/20/2022]
Abstract
LGI1 mutations predispose to a rare epilepsy syndrome and when inactivated in mice leads to early onset seizures and premature death. Histopathology of the mature brain soon after birth shows cortical dysplasia in Lgi1 null mice with hypercellularity in the outer cortical layers. Here we show extensive gene expression changes in neuronal precursor cells from Lgi1 null mice compared with wild type mice. The most significantly dysregulated pathway involves canonical axon guidance signaling with multiple networks involved in cell movement, adhesion and invasion related to actin cytoskeleton reorganization. The Lgi1 null NPCs show increased cell motility in vitro compared with normal counterparts. Dysregulation of genes critical to cell movement/migration and critical transcription factors involved in early neuronal development is a prominent feature. These studies provide a critical mechanistic link to the observation of increased cellularity in the outer layers of the developing cortex in Lgi1 null mice.
Collapse
|
44
|
Vaithiyanathan M, Safa N, Melvin AT. FluoroCellTrack: An algorithm for automated analysis of high-throughput droplet microfluidic data. PLoS One 2019; 14:e0215337. [PMID: 31042738 PMCID: PMC6493727 DOI: 10.1371/journal.pone.0215337] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/29/2019] [Indexed: 12/21/2022] Open
Abstract
High-throughput droplet microfluidic devices with fluorescence detection systems provide several advantages over conventional end-point cytometric techniques due to their ability to isolate single cells and investigate complex intracellular dynamics. While there have been significant advances in the field of experimental droplet microfluidics, the development of complementary software tools has lagged. Existing quantification tools have limitations including interdependent hardware platforms or challenges analyzing a wide range of high-throughput droplet microfluidic data using a single algorithm. To address these issues, an all-in-one Python algorithm called FluoroCellTrack was developed and its wide-range utility was tested on three different applications including quantification of cellular response to drugs, droplet tracking, and intracellular fluorescence. The algorithm imports all images collected using bright field and fluorescence microscopy and analyzes them to extract useful information. Two parallel steps are performed where droplets are detected using a mathematical Circular Hough Transform (CHT) while single cells (or other contours) are detected by a series of steps defining respective color boundaries involving edge detection, dilation, and erosion. These feature detection steps are strengthened by segmentation and radius/area thresholding for precise detection and removal of false positives. Individually detected droplet and contour center maps are overlaid to obtain encapsulation information for further analyses. FluoroCellTrack demonstrates an average of a ~92-99% similarity with manual analysis and exhibits a significant reduction in analysis time of 30 min to analyze an entire cohort compared to 20 h required for manual quantification.
Collapse
Affiliation(s)
- Manibarathi Vaithiyanathan
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Nora Safa
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, Louisiana, United States of America
| | - Adam T Melvin
- Cain Department of Chemical Engineering, Louisiana State University, Baton Rouge, Louisiana, United States of America
| |
Collapse
|
45
|
Rahikainen R, Öhman T, Turkki P, Varjosalo M, Hytönen VP. Talin-mediated force transmission and talin rod domain unfolding independently regulate adhesion signaling. J Cell Sci 2019; 132:jcs226514. [PMID: 30837291 DOI: 10.1242/jcs.226514] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/26/2019] [Indexed: 12/29/2022] Open
Abstract
Talin protein is one of the key components in integrin-mediated adhesion complexes. Talins transmit mechanical forces between β-integrin and actin, and regulate adhesion complex composition and signaling through the force-regulated unfolding of talin rod domain. Using modified talin proteins, we demonstrate that these functions contribute to different cellular processes and can be dissected. The transmission of mechanical forces regulates adhesion complex composition and phosphotyrosine signaling even in the absence of the mechanically regulated talin rod subdomains. However, the presence of the rod subdomains and their mechanical activation are required for the reinforcement of the adhesion complex, cell polarization and migration. Talin rod domain unfolding was also found to be essential for the generation of cellular signaling anisotropy, since both insufficient and excess activity of the rod domain severely inhibited cell polarization. Utilizing proteomics tools, we identified adhesome components that are recruited and activated either in a talin rod-dependent manner or independently of the rod subdomains. This study clarifies the division of roles between the force-regulated unfolding of a talin protein (talin 1) and its function as a physical linker between integrins and the cytoskeleton.
Collapse
Affiliation(s)
- Rolle Rahikainen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere 33014, Finland
- Fimlab Laboratories, Tampere 33520, Finland
| | - Tiina Öhman
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
| | - Paula Turkki
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere 33014, Finland
- Fimlab Laboratories, Tampere 33520, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, Tampere 33014, Finland
- Fimlab Laboratories, Tampere 33520, Finland
| |
Collapse
|
46
|
Rothenberg KE, Scott DW, Christoforou N, Hoffman BD. Vinculin Force-Sensitive Dynamics at Focal Adhesions Enable Effective Directed Cell Migration. Biophys J 2019; 114:1680-1694. [PMID: 29642037 DOI: 10.1016/j.bpj.2018.02.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 01/03/2023] Open
Abstract
Cell migration is a complex process, requiring coordination of many subcellular processes including membrane protrusion, adhesion, and contractility. For efficient cell migration, cells must concurrently control both transmission of large forces through adhesion structures and translocation of the cell body via adhesion turnover. Although mechanical regulation of protein dynamics has been proposed to play a major role in force transmission during cell migration, the key proteins and their exact roles are not completely understood. Vinculin is an adhesion protein that mediates force-sensitive processes, such as adhesion assembly under cytoskeletal load. Here, we elucidate the mechanical regulation of vinculin dynamics. Specifically, we paired measurements of vinculin loads using a Förster resonance energy transfer-based tension sensor and vinculin dynamics using fluorescence recovery after photobleaching to measure force-sensitive protein dynamics in living cells. We find that vinculin adopts a variety of mechanical states at adhesions, and the relationship between vinculin load and vinculin dynamics can be altered by the inhibition of vinculin binding to talin or actin or reduction of cytoskeletal contractility. Furthermore, the force-stabilized state of vinculin required for the stabilization of membrane protrusions is unnecessary for random migration, but is required for directional migration along a substrate-bound cue. These data show that the force-sensitive dynamics of vinculin impact force transmission and enable the mechanical integration of subcellular processes. These results suggest that the regulation of force-sensitive protein dynamics may have an underappreciated role in many cellular processes.
Collapse
Affiliation(s)
| | - David W Scott
- Lineberger Comprehensive Cancer Center, UNC Chapel, Chapel Hill, North Carolina
| | | | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina.
| |
Collapse
|
47
|
Cóndor M, Mark C, Gerum RC, Grummel NC, Bauer A, García-Aznar JM, Fabry B. Breast Cancer Cells Adapt Contractile Forces to Overcome Steric Hindrance. Biophys J 2019; 116:1305-1312. [PMID: 30902366 PMCID: PMC6451061 DOI: 10.1016/j.bpj.2019.02.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/24/2018] [Accepted: 02/01/2019] [Indexed: 01/03/2023] Open
Abstract
Cell migration through the extracellular matrix is governed by the interplay between cell-generated propulsion forces, adhesion forces, and resisting forces arising from the steric hindrance of the matrix. Steric hindrance in turn depends on matrix porosity, matrix deformability, cell size, and cell deformability. In this study, we investigate how cells respond to changes in steric hindrance that arise from altered cell mechanical properties. Specifically, we measure traction forces, cell morphology, and invasiveness of MDA-MB 231 breast cancer cells in three-dimensional collagen gels. To modulate cell mechanical properties, we either decrease nuclear deformability by twofold overexpression of the nuclear protein lamin A or we introduce into the cells stiff polystyrene beads with a diameter larger than the average matrix pore size. Despite this increase of steric hindrance, we find that cell invasion is only marginally inhibited, as measured by the fraction of motile cells and the mean invasion depth. To compensate for increased steric hindrance, cells employ two alternative strategies. Cells with higher nuclear stiffness increase their force polarity, whereas cells with large beads increase their net contractility. Under both conditions, the collagen matrix surrounding the cells stiffens dramatically and carries increased strain energy, suggesting that increased force polarity and increased net contractility are functionally equivalent strategies for overcoming an increased steric hindrance.
Collapse
Affiliation(s)
- Mar Cóndor
- Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain.
| | - Christoph Mark
- Department of Physics, University of Erlangen, Erlangen, Germany
| | - Richard C Gerum
- Department of Physics, University of Erlangen, Erlangen, Germany
| | - Nadine C Grummel
- Department of Physics, University of Erlangen, Erlangen, Germany
| | - Andreas Bauer
- Department of Physics, University of Erlangen, Erlangen, Germany
| | - José M García-Aznar
- Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Ben Fabry
- Department of Physics, University of Erlangen, Erlangen, Germany
| |
Collapse
|
48
|
Lee EJ, Kang MK, Kim YH, Kim DY, Oh H, Kim SI, Oh SY, Kang YH. Dietary Chrysin Suppresses Formation of Actin Cytoskeleton and Focal Adhesion in AGE-Exposed Mesangial Cells and Diabetic Kidney: Role of Autophagy. Nutrients 2019; 11:E127. [PMID: 30634545 PMCID: PMC6705957 DOI: 10.3390/nu11010127] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/16/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced glycation end products (AGE) play a causative role in the development of aberrant phenotypes of intraglomerular mesangial cells, contributing to acute/chronic glomerulonephritis. The aim of this study was to explore mechanistic effects of the flavonoid chrysin present in bee propolis and herbs on actin dynamics, focal adhesion, and the migration of AGE-exposed mesangial cells. The in vitro study cultured human mesangial cells exposed to 33 mM glucose and 100 μg/mL AGE-bovine serum albumin (AGE-BSA) for up to 5 days in the absence and presence of 1⁻20 μM chrysin. The in vivo study employed db/db mice orally administrated for 10 weeks with 10 mg/kg chrysin. The presence of ≥10 μM chrysin attenuated mesangial F-actin induction and bundle formation enhanced by AGE. Chrysin reduced the mesangial induction of α-smooth muscle actin (α-SMA) by glucose, and diminished the tissue α-SMA level in diabetic kidneys, indicating its blockade of mesangial proliferation. The treatment of chrysin inhibited the activation of vinculin and paxillin and the induction of cortactin, ARP2/3, fascin-1, and Ena/VASP-like protein in AGE-exposed mesangial cells. Oral administration of chrysin diminished tissue levels of cortactin and fascin-1 elevated in diabetic mouse kidneys. Mesangial cell motility was enhanced by AGE, which was markedly attenuated by adding chrysin to cells. On the other hand, chrysin dampened the induction of autophagy-related genes of beclin-1, LC3 I/II, Atg3, and Atg7 in mesangial cells exposed to AGE and in diabetic kidneys. Furthermore, chrysin reduced the mTOR activation in AGE-exposed mesangial cells and diabetic kidneys. The induction of mesangial F-actin, cortactin, and fascin-1 by AGE was deterred by the inhibition of autophagy and mTOR. Thus, chrysin may encumber diabetes-associated formation of actin bundling and focal adhesion and mesangial cell motility through disturbing autophagy and mTOR pathway.
Collapse
Affiliation(s)
- Eun-Jung Lee
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Min-Kyung Kang
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Yun-Ho Kim
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Dong Yeon Kim
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Hyeongjoo Oh
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Soo-Il Kim
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Su Yeon Oh
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| | - Young-Hee Kang
- Department of Food and Nutrition, Hallym University, Chuncheon, Kangwon-do 24252, Korea.
| |
Collapse
|
49
|
Brauer PR, Kim JH, Ochoa HJ, Stratton ER, Black KM, Rosencrans W, Stacey E, Hagos EG. Krüppel-like factor 4 mediates cellular migration and invasion by altering RhoA activity. ACTA ACUST UNITED AC 2018; 24:1-10. [PMID: 29498307 DOI: 10.1080/15419061.2018.1444034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Kru¨ppel like factor 4 (KLF4) is a transcription factor that regulates genes related to differentiation and proliferation. KLF4 also plays a role in metastasis via epithelial to mesenchymal transition. Here, we investigate the function of Klf4 in migration and invasion using mouse embryonic fibroblasts and the RKO human colon cancer cell line. Compared to wild-type, cells lacking Klf4 exhibited increased migration-associated phenotypes. In addition, overexpression of Klf4 in Klf4-/- MEFs attenuated the presence of stress fibers to wild-type levels. An invasion assay suggested that lack of Klf4 resulted in increased invasive capacity. Finally, analysis of RhoA showed elevated RhoA activity in both RKO and MEF cells. Taken together, our results strongly support the novel role of KLF4 in a post-translational regulatory mechanism where KLF4 indirectly modulates the actin cytoskeleton morphology via activity of RhoA in order to inhibit cellular migration and invasion.
Collapse
Affiliation(s)
- Philip R Brauer
- a Department of Biology , Colgate University , Hamilton , NY , USA
| | - Jee Hun Kim
- a Department of Biology , Colgate University , Hamilton , NY , USA
| | - Humberto J Ochoa
- a Department of Biology , Colgate University , Hamilton , NY , USA.,b Center for Cancer Research, Lab of Cancer Biology and Genetics , National Cancer Institute , Bethesda , MD , USA
| | | | - Kathryn M Black
- a Department of Biology , Colgate University , Hamilton , NY , USA.,c School of Medicine , Tulane University , New Orleans , LA , USA
| | | | - Eliza Stacey
- a Department of Biology , Colgate University , Hamilton , NY , USA
| | - Engda G Hagos
- a Department of Biology , Colgate University , Hamilton , NY , USA
| |
Collapse
|
50
|
Abstract
It is increasingly clear that mechanotransduction pathways play important roles in regulating fundamental cellular functions. Of the basic mechanical functions, the determination of cellular morphology is critical. Cells typically use many mechanosensitive steps and different cell states to achieve a polarized shape through repeated testing of the microenvironment. Indeed, morphology is determined by the microenvironment through periodic activation of motility, mechanotesting, and mechanoresponse functions by hormones, internal clocks, and receptor tyrosine kinases. Patterned substrates and controlled environments with defined rigidities limit the range of cell behavior and influence cell state decisions and are thus very useful for studying these steps. The recently defined rigidity sensing process provides a good example of how cells repeatedly test their microenvironment and is also linked to cancer. In general, aberrant extracellular matrix mechanosensing is associated with numerous conditions, including cardiovascular disease, aging, and fibrosis, that correlate with changes in tissue morphology and matrix composition. Hence, detailed descriptions of the steps involved in sensing and responding to the microenvironment are needed to better understand both the mechanisms of tissue homeostasis and the pathomechanisms of human disease.
Collapse
Affiliation(s)
- Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel 31096;
| | - Bo Yang
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore;
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; .,Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|